首页> 外文期刊>Journal of neuroinflammation >CXCL12 in astrocytes contributes to bone cancer pain through CXCR4-mediated neuronal sensitization and glial activation in rat spinal cord
【24h】

CXCL12 in astrocytes contributes to bone cancer pain through CXCR4-mediated neuronal sensitization and glial activation in rat spinal cord

机译:星形胶质细胞中的CXCL12通过CXCR4介导的大鼠脊髓神经元敏化和神经胶质活化而导致骨癌疼痛

获取原文
获取外文期刊封面目录资料

摘要

Background Previous studies have demonstrated that chemokine CXCL12 and its receptor CXCR4 are critical for pain sensitization, but the mechanisms involved are not clear. In this study, we investigated the specific cellular mechanisms of CXCL12/CXCR4 chemokine signaling in the development and maintenance of bone cancer pain after tumor cell implantation (TCI). Methods TCI in the tibial cavity of rats was used to establish a bone cancer pain model. Mechanical allodynia and thermal hyperalgesia were determined by measuring the paw withdrawal threshold and latency, respectively. The protein expression and cellular localization of CXCL12 and CXCR4 were detected by western blot and immunofluorescence staining. The sensitization of neurons, activation of astrocytes and microglia were examined by observing the immunofluorescence intensity of c-Fos, GFAP and IBA1. Results Our results demonstrated that CXCL12 was upregulated in a time-related manner, both in the dorsal root ganglia and spinal cord after TCI. Spinal CXCL12 was predominately expressed in astrocytes, and an intrathecal injection of astrocyte metabolic inhibitor fluorocitrate or selective JNK inhibitor SP600125 abolished TCI-induced CXCL12 production. A single intrathecal injection of a CXCL12 neutralizing antibody (10 μg/10 μl) at day 10 after TCI transiently reversed bone cancer pain in a dose-dependent manner. Whereas repetitive intrathecal administration of a CXCL12 neutralizing antibody (10 μg/10 μl, once a day from day 3 to 5 after TCI) significantly delayed the onset of TCI-induced pain behaviors for nearly five days. Spinal CXCR4 was also upregulated after TCI and colocalized with neurons, astrocytes and microglia. Blocking CXCR4 suppressed TCI-induced activation of neurons, astrocytes and microglia in the spinal cord at day 14. Repeated intrathecal administration of AMD3100 (5 μg/10 μl, once a day for three days) significantly delayed and suppressed the initiation and persistence of bone cancer pain in the early phase (at day 5, 6 and 7 after TCI) and in the late phase (at day 12, 13 and 14 after TCI) of bone cancer, respectively. Conclusions Taken together, these results demonstrate that CXCL12/CXCR4 signaling contributed to the development and maintenance of bone cancer pain via sensitizing neurons and activating astrocytes and microglia. Additionally, this chemokine signaling may be a potential target for treating bone cancer pain.
机译:背景技术先前的研究表明趋化因子CXCL12及其受体CXCR4对于疼痛敏化至关重要,但尚不清楚其机制。在这项研究中,我们研究了肿瘤细胞植入(TCI)后CXCL12 / CXCR4趋化因子信号传导在骨癌疼痛发生和维持中的特定细胞机制。方法采用大鼠胫骨腔TCI建立骨癌疼痛模型。机械性异常性疼痛和热痛觉过敏分别通过测量爪缩回阈值和潜伏期来确定。 Western blot和免疫荧光染色检测CXCL12和CXCR4的蛋白表达和细胞定位。通过观察c-Fos,GFAP和IBA1的免疫荧光强度来检查神经元的敏化,星形胶质细胞和小胶质细胞的活化。结果我们的结果表明,TCI后,背根神经节和脊髓中CXCL12均呈时间相关上调。脊髓CXCL12主要在星形胶质细胞中表达,鞘内注射星形胶质细胞代谢抑制剂氟柠檬酸盐或选择性JNK抑制剂SP600125消除了TCI诱导的CXCL12的产生。在TCI后第10天,鞘内注射CXCL12中和抗体(10μg/ 10μl)以剂量依赖性方式暂时逆转骨癌疼痛。而鞘内注射CXCL12中和抗体(10μg/ 10μl,在TCI后第3天到第5天每天一次)显着延迟了TCI诱导的疼痛行为发作近五天。 TCI后脊髓CXCR4也上调,并与神经元,星形胶质细胞和小胶质细胞共定位。在第14天,阻断CXCR4抑制了TCI诱导的脊髓中神经元,星形胶质细胞和小胶质细胞的活化。重复鞘内施用AMD3100(5μg/ 10μl,每天一次,持续三天)显着延迟并抑制了骨的起始和持久性在骨癌的早期(TCI后的第5、6和7天)和晚期(TCI后的第12、13和14天)患癌疼痛。结论综上所述,这些结果表明CXCL12 / CXCR4信号通过敏化神经元,激活星形胶质细胞和小胶质细胞而有助于骨癌疼痛的发展和维持。另外,该趋化因子信号传导可能是治疗骨癌疼痛的潜在靶标。

著录项

相似文献

  • 外文文献
  • 中文文献
  • 专利
获取原文

客服邮箱:kefu@zhangqiaokeyan.com

京公网安备:11010802029741号 ICP备案号:京ICP备15016152号-6 六维联合信息科技 (北京) 有限公司©版权所有
  • 客服微信

  • 服务号